ArticlePDF Available

Stimuli-Responsive Polymer-Prodrug Hybrid Nanoplatform for Multistage siRNA Delivery and Combination Cancer Therapy

Authors:

Abstract and Figures

Nanoparticles (NPs) formulated with cationic lipids and/or polymers have shown substantial potential for systemic delivery of RNA therapeutics such as small interfering RNA (siRNA) for the treatment of cancer and other diseases. While both cationic lipids and polymers have demonstrated the promise to facilitate siRNA encapsulation and endosomal escape, they could also hamper cytosolic siRNA release due to charge interaction and induce potential toxicities. Herein, a unique polymer-prodrug hybrid NP platform was developed for multistage siRNA delivery and combination cancer therapy. This NP system is composed of (i) a hydrophilic polyethylene glycol (PEG) shell, (ii) a hydrophobic NP core made with a tumor microenvironment (TME) pH-responsive polymer, and (iii) charge-mediated complexes of siRNA and amphiphilic cationic mitoxantrone (MTO)-based prodrug that are encapsulated in the NP core. After intravenous administration, the long-circulating NPs accumulate in tumor tissues and then rapidly release the siRNA-prodrug complexes via TME pH-mediated NP disassociation for subsequent tissue penetration and cytosolic transport. With the over-expressed esterase in tumor cells to hydrolyze the amphiphilic structure of the prodrug and thereby induce destabilization of the siRNA-prodrug complexes, the therapeutic siRNA and anticancer drug MTO can be efficiently released in the cytoplasm, ultimately leading to the combinational inhibition of tumor growth via concurrent RNAi-mediated gene silencing and MTO-mediated chemotherapy.
Content may be subject to copyright.
Stimuli-Responsive PolymerProdrug Hybrid Nanoplatform for
Multistage siRNA Delivery and Combination Cancer Therapy
Phei Er Saw,
,
Herui Yao,
,
Chunhao Lin,
,
Wei Tao,
§
Omid C Farokhzad,*
,§
and Xiaoding Xu*
,,
Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun
Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
RNA Biomedical Institute, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
§
Center for Nanomedicine and Department of Anesthesiology, Brigham and Womens Hospital, Harvard Medical School, Boston,
Massachusetts 02115, United States
*
SSupporting Information
ABSTRACT: Nanoparticles (NPs) formulated with cationic lipids and/or
polymers have shown substantial potential for systemic delivery of RNA
therapeutics such as small interfering RNA (siRNA) for the treatment of cancer
and other diseases. While both cationic lipids and polymers have demonstrated the
promise to facilitate siRNA encapsulation and endosomal escape, they could also
hamper cytosolic siRNA release due to charge interaction and induce potential
toxicities. Herein, a unique polymerprodrug hybrid NP platform was developed
for multistage siRNA delivery and combination cancer therapy. This NP system is
composed of (i) a hydrophilic polyethylene glycol (PEG) shell, (ii) a hydrophobic
NP core made with a tumor microenvironment (TME) pH-responsive polymer,
and (iii) charge-mediated complexes of siRNA and amphiphilic cationic mitoxantrone (MTO)-based prodrug that are
encapsulated in the NP core. After intravenous administration, the long-circulating NPs accumulate in tumor tissues and then
rapidly release the siRNAprodrug complexes via TME pH-mediated NP disassociation for subsequent tissue penetration and
cytosolic transport. With the overexpressed esterase in tumor cells to hydrolyze the amphiphilic structure of the prodrug and
thereby induce destabilization of the siRNAprodrug complexes, the therapeutic siRNA and anticancer drug MTO can be
eciently released in the cytoplasm, ultimately leading to the combinational inhibition of tumor growth via concurrent RNAi-
mediated gene silencing and MTO-mediated chemotherapy.
KEYWORDS: Nanoparticles, stimuli-responsive, prodrug, multistage siRNA delivery, combination cancer therapy
RNA interference (RNAi) technology has demonstrated
tremendous potential for disease treatment by specic
silencing the expression of target gene(s), especially those
encoding undruggableproteins.
13
However, RNAi ther-
apeutics such as small interfering RNA (siRNA) are susceptible
to nucleases and cannot readily cross cell membrane due to
their polyanionic and biomacromolecular characteristics.
46
Therefore, specic delivery carriers are required to protect
siRNA from enzymatic degradation and to facilitate its cellular
uptake and cytosolic transport. The most commonly used
carriers are cationic lipids and polymers,
712
which can bind
siRNA to form complexes (lipoplexes or polyplexes) via
electrostatic interaction. While both cationic lipids and
polymers can improve the siRNA stability, increase its cellular
uptake, and enhance endosomal escape,
1316
they could also
hamper the cytosolic siRNA release due to charge
interaction.
1719
More importantly, due to the lower charge
density and molecular weight of siRNA compared to plasmid
DNA, high doses of cationic lipids or polymers are usually used
to increase the siRNA loading ability and stability,
20,21
which in
turn causes severe cytotoxicity to normal tissues.
In the past decade, much eort has been paid to develop safe
and eective siRNA delivery systems. To date, various
innovative approaches, such as the use of coordination
chemistry
2124
and a guanidine group
15,2528
with stronger
RNA binding anity, have been employed to construct siRNA
delivery carries. Nevertheless, stronger RNA binding anity in
these strategies may also hamper the cytosolic siRNA release.
Although the use of bioresponsive linkages (e.g., disulde
bond) can endow the carriers with stimuli-responsive
biodegradability,
12,2932
the generated small cationic molecules
after degradation may still induce potential toxicity and side
eects.
To address these issues, we herein developed a tumor
microenvironment (TME)-responsive polymerprodrug hy-
brid nanoparticle (NP) platform for multistage siRNA delivery
and combination cancer therapy. Prodrug is a promising
strategy to improve the selectivity and ecacy of a chemo-
Received: April 22, 2019
Revised: August 5, 2019
Published: August 5, 2019
Letter
pubs.acs.org/NanoLett
Cite This: Nano Lett. 2019, 19, 59675974
© 2019 American Chemical Society 5967 DOI: 10.1021/acs.nanolett.9b01660
Nano Lett. 2019, 19, 59675974
Downloaded via SUN YAT SEN (ZHONGSHAN) UNIV on March 16, 2020 at 08:07:50 (UTC).
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.
therapeutic drug.
33,34
By modifying chemotherapeutic drugs
with dierent ligands, the prodrugs show high stability, long
blood circulation, low immunogenicity, and specic targeting
to tissues or cells.
3537
More importantly, several anticancer
drugs such as mitoxantrone (MTO) and metformin possess
intrinsic cationic characteristic.
28,38,39
Therefore, rational
modication of these drugs may provide valuable opportunity
to form electrostatic interaction with the negatively charged
siRNA, which not only avoids the potential toxicity of
traditional cationic lipids and polymers but also provides the
feasibility to achieve combination cancer therapy. To this end,
we chose MTO and synthesized an amphiphilic cationic MTO-
based prodrug (Scheme 1A), which can coassemble with a
TME pH-responsive polymer methoxyl-poly(ethylene glycol)-
b-poly(2-(pentamethyleneimino)ethyl methacrylate) (Meo-
PEG-b-PPMEMA)
40,41
to form polymerprodrug hybrid
NPs. After siRNA encapsulation, the resulting NP platform
shows the following features (Scheme 1B): (i) hydrophilic
PEG chains prolong blood circulation and thereby enhance
tumor accumulation via the enhanced permeability and
retention (EPR) eect; (ii) TME pH-triggered protonation
of the hydrophobic PPMEMA segment induces the rapid NP
disassociation and exposure of siRNAprodrug complexes that
can penetrate tumor tissues and enhance cytosolic siRNA
transport; (iii) overexpressed esterase in the tumor cells can
hydrolyze the amphiphilic structure of prodrug and thus
induce destabilization of the siRNAprodrug complexes,
4244
leading to ecient cytosolic release of siRNA and intact MTO
to accomplish combination cancer therapy.
In this work, Polo-like kinase 1 (PLK1) was chosen as a
therapeutic target and we systematically evaluated the TME
pH-responsive polymerprodrug hybrid NPs for PLK1 siRNA
(siPLK1) delivery and their anticancer ecacy. PLK1 is a
proto-oncogene that overexpressed in several dierent cancers
(e.g., breast cancer and prostate cancer)
4547
and its high
expression is correlated with the low survival rate of
patients.
4851
Previous studies have demonstrated that PLK1
not only plays an important role in regulating cell mitosis but
also can reduce the therapeutic ecacy of chemotherapeutic
drugs via microtubule rearrangement and DNA damage
recovery.
5254
Therefore, concurrent down-regulation of
PLK1 expression is expected to improve the therapeutic
ecacy of cancer chemotherapy. Our in vivo results
demonstrate that the TME pH-responsive hybrid NP platform
can eciently deliver siPLK1 to tumor cells, leading to
combinational inhibition of tumor growth of breast cancer via
concurrent PLK1 silencing and MTO-based chemotherapy.
Results and Discussion. The structure of the MTO-based
prodrug is shown Scheme 1A. MTO is a clinically approved
anticancer drug and has been widely used for the treatment of
acute myelogenous leukemia, breast cancer, and advanced
prostate cancer.
55
Compared to other anticancer drugs (e.g.,
cisplatin and docetaxel), it is convenient to track the MTO
distribution in real-time both in vitro and in vivo, due to its NIR
characteristic with excitation at 610 and 660 nm and emission
at 685 nm.
5658
More importantly, the cationic nature of MTO
may provide the possibility to complex negatively charged
siRNA for combination cancer therapy. Therefore, to mimic
the typical structure of cationic lipids,
7
two hydrophobic tails
were conjugated to the MTO structure via ester bond to obtain
an amphiphilic cationic MTO-based prodrug (denoted SA-
MTO). Successful synthesis of the prodrug SA-MTO was
conrmed by nuclear magnetic resonance (NMR, Figure S1).
Fluorescence spectroscopy analysis shows that the incorpo-
ration of hydrophobic tails does not aect the NIR
characteristic (Figure 1A), ensuring the feasibility of real-
time tracking the MTO distribution. We rst employed high-
performance liquid chromatograph (HPLC) to examine
whether the prodrug can be hydrolyzed by esterase to release
the pharmacologically active MTO. As shown in Figure 1B, the
typical signal corresponding to the intact MTO molecules can
be observed when incubating the prodrug with esterase for 4 h.
By prolonging the incubation time, more intact MTO
molecules can be detected and almost all the prodrug
molecules have been hydrolyzed 12 h later, which could
ensure the pharmacological activity of MTO since esterase has
been demonstrated to be overexpressed in cancer cells.
4244
After the successful synthesis of the prodrug and validation
of its esterase-triggered hydrolysis, we next synthesized the
TME pH-responsive polymer Meo-PEG-b-PPMEMA (pKa
6.89) (Figures S2 and S3),
40,41
which was subsequently
formulated with the prodrug SA-MTO to prepare the
polymerprodrug hybrid NPs. As shown in Figure 1C, when
mixing the dimethylformamide (DMF) mixture of the prodrug
SA-MTO and polymer Meo-PEG-b-PPMEMA with the siRNA
aqueous solution followed by adding to deionized (DI) water,
self-assembled spherical siRNA-loaded NPs can be formed. In
this polymerprodrug hybrid NP system, the amphiphilic
cationic prodrug SA-MTO rst forms complexes the negatively
Scheme 1. (A) Chemical Structure of the Amphiphilic
Cationic Prodrug SA-MTO and TME pH-Responsive
Polymer Meo-PEG-b-PPMEMA; (B) Schematic Illustration
of the TME pH-Responsive PolymerProdrug Hybrid
Nanoplatform for Multistage siRNA Delivery and
Combination Cancer Therapy
a
a
After intravenous injection, the hybrid NPs can extravasate from
leaky tumor vasculature and accumulate in the tumor tissue (a).
Subsequently, the TME pH-triggered NP disassociation induces rapid
release of the siRNAprodrug complexes (b), which can penetrate
tumor tissue and enter the tumor cells (c). After cellular uptake (d),
the overexpressed esterase in tumor cells can hydrolyze the ester bond
of the prodrug SA-MTO (e), leading to ecient release of therapeutic
siRNA and intact MTO in the cytoplasm, which thereby inhibit the
tumor growth via concurrent RNAi therapy (f) and chemotherapy
(g).
Nano Letters Letter
DOI: 10.1021/acs.nanolett.9b01660
Nano Lett. 2019, 19, 59675974
5968
charged siRNA in the DMF solution via electrostatic
interaction with the hydrophobic tails of the prodrug
positioned on the surface of the complexes. When adding
these complexes to DI water, they can be embedded in the
hydrophobic cores of the self-assembled polymer Meo-PEG-b-
PPMEMA via the hydrophobic interaction with PPMEMA
segment.
31,5961
We varied the N/P molar ratio by changing
the prodrug amount to adjust the physiochemical properties of
the siRNA-loaded NPs. As shown in Figure 1D, as the N/P
ratio increases from 5 to 30, the resulting NPs (denoted NP5,
NP10, NP15, NP20, and NP30) show increased siRNA
encapsulation eciency (EE) from 35.1% to 68.9% and
particle size from 70 to 142 nm. One possible reason is that
the increased N/P ratio induces much stronger electrostatic
interaction with the siRNA, leading to encapsulating more
siRNAprodrug complexes in the NPs with larger particle size.
Similarly, the ζpotential and drug loading level (LL) of the
resulting NPs increases from NP5 to NP30 as the increased
amount of the prodrug SA-MTO was used.
Since the TME pH-responsive polymer Meo-PEG-b-
PPMEMA was incorporated into the NPs, we next examined
the TME pH response of these NPs. Taking NP15 for
example, when this nanoplatform was incubated in the solution
at a pH below the pKaof the polymer (e.g., pH 6.8), the rapid
protonation of the polymer Meo-PEG-b-PPMEMA induced a
rapid NP dissociation and led to a dramatic decrease in NP
number (Figure 1E). Furthermore, unlike the well-dened NPs
with spherical morphology at pH 7.4, small size particles and
large amorphous aggregates can be observed in the pH 6.8
solution, which is further proven by dynamic light scattering
(DLS) analysis (Figure S4) and may correspond to the
exposed siRNAprodrug complexes and ionized polymer,
respectively. To further conrm the presence of siRNA
prodrug complexes after NP dissociation, siRNA labeled with
uorescein (FL) and its quencher (Dabcyl) were encapsulated
in the NPs and the FL uorescence at dierent pHs was
examined.
15,62,63
The naked siRNA can be degraded by RNase
and thus FL and its quencher are separated, leading to a
signicant increase in the FL uorescence (Figure S5). In
contrast, because the cationic prodrug SA-MTO is able to
condense siRNA to form stable complexes that could protect
siRNA from degradation, no obvious change can be seen in the
FL uorescence although NP dissociation appears at pH 6.8.
Furthermore, this TME pH-triggered rapid NP dissociation
results in a fast siRNA release. As shown in Figure 1F, more
than 60% of the loaded siRNA has been released within 8 h at
pH 6.8, while only around 10% of the loaded siRNA is released
at pH 7.4. Moreover, because the addition of esterase can
hydrolyze the ester bond in the cationic prodrug SA-MTO and
subsequently induce destabilization of the siRNAprodrug
complexes, the NPs show faster siRNA release than that of the
NPs incubated in the pH 6.8 solution without esterase.
Notably, because the NPs can maintain their structure in the
pH 7.4 solution and prevent the esterase from hydrolyzing the
prodrug SA-MTO encapsulated in the hydrophobic cores,
there is no signicant dierence in the siRNA release
compared to the NPs incubated in the pH 7.4 solution
without esterase. Similar results can be found in the MTO
release prole (Figure 1G). In the presence of esterase, the
NPs incubated in the pH 6.8 solution show faster MTO release
than that incubated in the pH 7.4 solution. In the absence of
esterase, there is nearly no dierence in the free MTO release
Figure 1. (A) Fluorescence emission spectra and uorescence images of the prodrug SA-MTO and free MTO in the mixture of DMF and
deionized water (8/2, v/v). (B) HPLC proles of the prodrug SA-MTO and free MTO incubated with esterase for dierent times. (C) Size
distribution and morphology of the NP15 in aqueous solution at pH 7.4. (D) Size, zeta potential (ζ), siRNA encapsulation eciency (EE%), and
prodrug loading level (LL%) of the siRNA-loaded NPs prepared at dierent N/P molar ratios. (E) NP number (count rate) of the NP15 in PBS
buer at pH 6.8 for dierent times and the morphology of the NP15 after incubating in PBS buer at pH 6.8 for 10 min. (F, G) Cumulative siRNA
(F) and free MTO (G) release from the NP15 incubated in PBS buer at dierent pHs with or without esterase.
Nano Letters Letter
DOI: 10.1021/acs.nanolett.9b01660
Nano Lett. 2019, 19, 59675974
5969
between these two pHs even the appearance of NP
dissociation at pH 6.8.
We next investigated whether the TME pH-triggered NP
dissociation can enhance the siRNA uptake and improve gene
silencing ecacy. Luciferase-expressing human breast cancer
cells (Luc-MDA-MB-231) were used to incubate with Cy3-
labeled siRNA-loaded NP15 at dierent pHs for 2 h, and the
siRNA uptake was viewed by a confocal laser-scanning
microscope (CLSM). As shown in Figure 2, the cells show
stronger siRNA uptake at pH 6.8 (Figure 2Ba) than pH 7.4
(Figure 2Aa). Noteworthy, these internalized siRNA molecules
at pH 6.8 are mainly distributed in the cytoplasm, as
demonstrated by colocalization analysis of endosomes and
siRNA molecules (Figure 2Bf). In addition, with the abundant
intracellular esterase to hydrolyze the ester bond of the cationic
prodrug SA-MTO and subsequently induce destabilization of
the siRNAprodrug complexes, the encapsulated MTO
molecules can be released (Figure 2Be) and enter the nuclei
(Figure 2Bh). The similar result can be found in the cells
treated with the siRNAprodrug complexes (Figure S6),
indicating that the improved cellular uptake and endosomal
escape is mainly due to the exposed siRNAprodrug
complexes after TME pH-triggered NP dissociation. The
improved siRNA uptake at pH 6.8 is further quantied by ow
cytometry analysis (Figure 2C,D), in which the siRNA uptake
at pH 6.8 is around 5-fold higher than that of the cells treated
with the siRNA-loaded NPs at pH 7.4.
Having conrmed the improved siRNA uptake induced by
the TME pH-triggered NP dissociation, we then examined the
gene silence ecacy by encapsulating luciferase siRNA (siLuc)
into the NPs. As shown in Figure 2E, all the siLuc-loaded NPs
can down-regulate Luc expression at a 20 nM siRNA dose,
though the silencing ecacy diers depending on the NP
formulation. Moreover, with the improved siRNA uptake at
pH 6.8 (Figure 2Ba,C), the siLuc-loaded NPs oer much
better gene silencing ecacy than that at pH 7.4. Especially for
the NP15 platform, its gene silencing ecacy is close to the
commercial Lipofectamine 2000 (Lipo2K) at pH 6.8. By
replacing the polymer Meo-PEG-b-PPMEMA with non-pH-
responsive polymer methoxyl-poly(ethylene glycol)-b-poly-
(lactic-co-glycolic acid) (Meo-PEG-b-PLGA), there is nearly
no dierence in the gene silencing ecacy between pH 7.4 and
6.8, highlighting the importance of TME pH-triggered NP
dissociation to improve the gene silencing. In comparison, the
siLuc-loaded NP15, NP20, and NP30 with higher siRNA
encapsulation eciency show better silencing ecacy than that
of other NP platforms (NP5 and NP10) with relatively lower
siRNA encapsulation eciency. Herein, we chose NP15 for the
following experiments since this NP platform shows moderate
ζpotential and smaller particle size (<100 nm).
Figure 2. (A, B) CLSM images of Luc-MDA-MB-231 cells incubated
with the Cy3-labeled siLuc-loaded NP15 at pH 7.4 (A) or 6.8 (B) for
2 h. The endosomes and nuclei were stained with lysotracker green
and Hoechst 33342, respectively. (C, D) Flow cytometry prole (C)
and mean uorescence intensity (MFI, D) of Luc-MDA-MB-231 cells
incubated with the Cy3-labeled siLuc-loaded NP15 at dierent pHs
for 2 h. (E) Luc expression in Luc-MDA-MB-231 cells treated with
the Lipo2K/siLuc complexes or the siLuc-loaded NPs at a 20 nM
siRNA dose. For each NP formulation, the NPs loaded scrambled
siRNA were used as control.
Figure 3. (A, B) Western blot analysis of PLK1 expression in MDA-
MB-231 cells treated with the siPLK1-loaded NP15 at pH 7.4 (A) or
6.8 (B). (C) Immunouorescence analysis of PLK1 expression (red
uorescence) in MDA-MB-231 cells treated with the siPLK1-loaded
NP15 at a 20 nM siRNA dose. White arrows indicate the
unconsolidated nuclei. (D, E) Flow cytometry analysis (D) and
quantication of apoptosis (E) of MDA-MB-231 cells treated with
siLuc- or siPLK1-loaded NP15 at a 20 nM siRNA dose. (F)
Proliferation prole of MDA-MB-231 cells treated with siLuc- or
siPLK1-loaded NP15 at a 20 nM siRNA dose. The cells incubated
with the blank NPs were used as the control in these experiments.
Nano Letters Letter
DOI: 10.1021/acs.nanolett.9b01660
Nano Lett. 2019, 19, 59675974
5970
After NP screening to obtain the optimal polymerprodrug
hybrid NP platform (NP15), we next examined the feasibility
of using this RNAi nanoplatform to silence PLK1, a proto-
oncogene that overexpressed in several dierent cancers
including breast cancer.
46,47,49
As shown in Figure 3A, the
siPLK1-loaded NP15 can down-regulate PLK1 expression in
the MDA-MB-231 cells at pH 7.4 and around 70% knockdown
can be achieved at a 20 nM siRNA dose (Figure S7). More
importantly, due to the improved siRNA uptake at pH 6.8, the
siPLK1-loaded NPs exhibit stronger PLK1 silencing and
around 90% of the PLK1 expression is suppressed at a 20
nM siRNA dose (Figure 3B, Figure S7). Immunouorescence
staining analysis (Figure 3C) also indicates similar ndings.
Compared to the residual PLK1 (red uorescence) in the cells
treated with the siPLK1-loaded NP15 at pH 7.4, red
uorescence is nearly absent in the cells treated with the
same NPs at pH 6.8. Due to this ecient PLK1 silencing, the
nuclei become shrunk and even unconsolidated (white arrow
in Figure 3), implying the presence of cell apoptosis or
necrosis. The ow cytometry analysis further proves our
prediction (Figure 3D,E). For the cells treated with the
siPLK1-loaded NP15 at pH 6.8, the percentage of apoptotic
cells reaches around 60%, which is 3- or 2-fold higher than that
of the cells treated with the siLuc- (22.5%) or siPLK1-loaded
NP15 (38.6%) at pH 7.4, respectively. In addition, the PLK1
silencing also signicantly inhibits the proliferation of MDA-
MB-231 cells. As shown in Figure 3F, after 24 h incubation
with the siPLK1-loaded NP15 at pH 6.8 followed by
incubation with fresh culture medium for 6 days, only half of
the cells are alive. However, within the same time frame, there
is about 8- or 4-fold increase in the number of cells treated
with the siLuc- or siPLK1-loaded NP15 at pH 7.4, respectively.
Notably, compared to the cells treated the blank NPs with no
payload (Control), the observed apoptosis (Figure 3D,E) and
inhibited proliferation of the cells treated with the siLuc-loaded
NP15 is mainly due to the loaded SA-MTO prodrug in the
NPs,
57
but not the toxicity of the polymer Meo-PEG-b-
PPMEMA (Figure S8, around 90% cell viability at a polymer
concentration of 100 mg/L). According to the drug loading
level and NP formulation shown in Figure 1D, the nal
concentration of MTO and the polymer Meo-PEG-b-
PPMEMA is 1.8 and 80 mg/L, respectively, when 20 nM
siRNA dose is used in the in vitro experiments (Figure 3CF).
Having validated the ecient gene silencing of the hybrid
NP15 platform in vitro,wenextexamineditsin vivo
pharmacokinetics (PK) and biodistribution (BioD). In general,
uorescent dyes are used to label NPs to facilitate real-time
monitoring of their circulation and distribution in vivo.
However, the use of uorescent dyes may also aect the
physiochemical properties of the resulting NPs and thus the
obtained results may not reect the real in vivo behaviors of the
original NPs. Herein, due to the NIR characteristic of MTO,
no uorescent dye was used to label the hybrid NP15 platform,
which can eciently avoid the complicated process for dye-
labeling and its potential inuence on the physiochemical
properties of the resulting NPs. Figure 4A shows the blood
circulation prole of the siLuc-loaded NP15 after intravenous
(iv) injection to healthy mice (1 nmol of siRNA dose per
mouse, 4.5 mg/kg MTO-equivalent dose, n= 3). Due to the
protection by PEG outer layer,
6466
the siRNA-loaded NPs
Figure 4. (A) Blood circulation prole of naked siRNA, free MTO, and the siLuc-loaded NP15. (B) Overlaid uorescence image of the MDA-MB-
231 xenograft tumor-bearing nude mice at 24 h post injection of naked siRNA, free MTO, and the siLuc-loaded NP15. Tumors are indicated by
ellipses. (C) Biodistribution of naked siRNA, free MTO and the siLuc-loaded NP15 in the tumors and major organs of the MDA-MB-231
xenograft tumor-bearing nude mice sacriced at 24 h postinjection. (D) CLSM images of the 3D tumor spheroids incubated with the siLuc-loaded
NP15 and PLGA NPs (Control NPs) at pH 7.4 or 6.8 for 4 h. (E) Fluorescence images of the MDA-MB-231 tumor sections at 4 h postinjection of
naked siRNA, free MTO, and siLuc-loadedNP15 and PLGA NPs (Control NPs).
Nano Letters Letter
DOI: 10.1021/acs.nanolett.9b01660
Nano Lett. 2019, 19, 59675974
5971
show long blood circulation with a half-life of around 1.02 h. In
contrast, naked siRNA (1 nmol of siRNA dose per mouse) or
free MTO (4.5 mg/kg) is cleared rapidly from the blood and
their blood circulation half-lives are less than 10 min. The
BioD was examined by iv injection of siLuc-loaded NP15 to
the MDA-MB-231 xenograft tumor-bearing mice (1 nmol of
siRNA dose per mouse, 4.5 mg/kg MTO-equivalent dose, n=
3). Figure 4B shows the whole body uorescence image of the
mice at 24 h post injection. The siRNA-loaded NPs show a
much higher tumor accumulation than naked siRNA or free
MTO. The tumors and major organs (heart, liver, spleen, lung,
and kidney) were harvested (Figure S9) and the BioD was
determined by examining the uorescence intensity of each
tissue. As shown in Figure 4C, the siRNA-loaded NPs show
more than 7-fold higher tumor accumulation than that of
naked siRNA or free MTO.
With the above promising BioD result, we subsequently
investigated whether the tumor-accumulated NPs can respond
to TME pH to enhance tumor penetration. The 3D tumor
spheroids were rst constructed to evaluate the penetration
ability.
67,68
From the CLSM images shown in Figure 4D, red
uorescence corresponding to the siRNA-loaded NPs is mainly
located on the periphery of the 3D tumor spheroid treated
with the siLuc-loaded NP15 at pH 7.4 for 4 h. In contrast, for
the spheroid treated with the same NPs at pH 6.8, due to the
TME pH-triggered NP disassociation to expose the small size
siRNAprodrug complexes with strong tissue penetration
ability, red uorescence can be clearly observed in the interior
area of the 3D tumor spheroid. This result is similar to that of
the 3D tumor spheroid incubated with the siRNAprodrug
complexes (Figure S10). By replacing the TME pH-responsive
polymer with the Meo-PEG-b-PLGA polymer (denoted
control NPs), there is no obvious dierence in the NP
distribution between pH 7.4 and 6.8. To further demonstrate
the improved tumor penetration induced by the TME pH-
triggered NP disassociation, the MDA-MB-231 xenograft
tumor-bearing mice received the iv injection of the siLuc-
loaded NP15, and tumors were collected at 4 h postinjection
and sectioned for CD31 staining.
14,58,69
As shown in Figure 4E,
the exposed siRNAprodrug complexes from the siRNA-
loaded NPs can extravasate from tumor vessels and deeply
penetrate the extravascular tumor parenchyma. In contrast, due
to the absence of TME pH response, most of the control NPs
(red uorescence) can be seen in or around the tumor vessels,
with only a small fraction entering the extravascular tumor
parenchyma.
We nally evaluated the in vivo PLK1 silencing ecacy and
anticancer eect of the polymerprodrug hybrid NP platform.
To examine the in vivo PLK1 silencing, the siPLK1-loaded
NP15 was intravenously injected to the MDA-MB-231
xenograft tumor-bearing mice (1 nmol of siRNA dose per
mouse, 4.5 mg/kg MTO-equivalent dose, n= 3) for three
consecutive days. As shown in Figure 5A,B, the administration
of the siPLK1-loaded NPs induces 70% knockdown in the
PLK1 expression. The immunohistochemistry (IHC) staining
analysis also indicates the ecient PLK1 silencing of the
siPLK1-loaded NP15 (Figures 5C). With this promising in vivo
PLK1 silencing, we next examined anticancer eect by iv
injection of the siPLK1-loaded NP15 to the MDA-MB-231
xenograft tumor-bearing mice once every 2 days (1 nmol of
siRNA dose per mouse, 4.5 mg/kg MTO-equivalent dose, n=
5). As shown in Figure 5DF, after four consecutive injections,
Figure 5. (A)(C) Western blot (A, B) and IHC analysis (C) of PLK1 expression in the tumor tissues of the MDA-MB-231 xenograft tumor-
bearing nude mice treated with siLuc- (Control NPs) or siPLK1-loaded NP15. *** P< 0.001. (D, E) Tumor size (D) and weight (E) of the MDA-
MB-231 xenograft tumor-bearing nude mice treated with PBS, naked siPKK1, free MTO, and siLuc-, and siPLK1-loaded NP15. The intravenous
injections are indicated by the arrows. ** P< 0.01; *** P< 0.001. (F) Representative photograph of the MDA-MB-231 xenograft tumor-bearing
nude mice in each group at day 18. Tumors are indicated by ellipses. (G, H) TUNEL (G) and H&E (H) staining of the MDA-MB-231 tumor
tissues after systemic treatment in each group. TUNEL-positive apoptotic cells were stained with red uorescence.
Nano Letters Letter
DOI: 10.1021/acs.nanolett.9b01660
Nano Lett. 2019, 19, 59675974
5972
the siPLK1-loaded NPs can signicantly inhibit tumor growth
and there is less than 2-fold increase (from 69 to 137 mm3)
in tumor size within 18 days. In contrast, for the mice treated
with PBS, naked siPLK1 (1 nmol of siRNA dose per mouse),
or free MTO (4.5 mg/kg), they show more than 7-fold
increase in their tumor size and tumor weight. Due to the
encapsulation of the prodrug SA-MTO in the siLuc-loaded
NPs, they are also able to inhibit tumor growth (>4-fold
increase in the tumor size within 18 day evaluation period).
However, the tumor inhibition ecacy is lower than that of the
siPLK1-loaded NPs showing the characteristic of combination
therapy, i.e., RNAi therapy from siPLK1 and chemotherapy
from MTO. Histological analysis of tumor slides further
demonstrates the fact that the siPLK1-loaded NPs are the most
eective in inducing cell apoptosis (Figure 5G) and inhibiting
cell proliferation (Figure 5H). Moreover, the administration of
siPLK1-loaded NPs does not aect the mouse body weight
(Figure S11). Histological analysis (Figure S12) shows that
there are no obvious histological changes in the main organs
(heart, liver, spleen, lung, and kidney). To further assess the in
vivo safety, the siPLK1-loaded NP15 was administrated to the
healthy mice (1 nmol of siRNA dose per mouse, 4.5 mg/kg
MTO-equivalent dose, n= 3) via iv injection. Immune
response analysis shows that the level of representative
cytokines (TNF-α, IFN-γ, IL-6, and IL-12) is in the normal
range (Figure S13). Blood routine analysis indicates that
aspartate aminotransferase (AST), alanine aminotransferase
(ALT), alkaline phosphatase (ALKP), blood urine nitrogen
(BUN), creatinine, and total protein are in the normal range
(Figure S14). Taken together, these results demonstrate good
biocompatibility of our newly developed polymerprodrug
hybrid NP platform.
Conclusion. In summary, we have developed a new TME
pH-responsive polymerprodrug hybrid NP platform for
multistage siRNA delivery and combination cancer therapy.
This long-circulating NP platform can rst accumulate in the
tumor tissues and then rapidly respond to TME pH to expose
the small size siRNAprodrug complexes, which subsequently
penetrate tumor tissues and enter the tumor cells. With the
overexpressed esterase in the tumor cells to break the prodrug
structure, the therapeutic siRNA and anticancer drug can be
eciently released in the cytoplasm, inducing combinational
inhibition of tumor growth via concurrent RNAi therapy from
siPLK1 and chemotherapy from MTO. This hybrid NP
platform could be used as an eective vehicle for the systemic
delivery of various biomacromolecules (e.g., nucleic acids and
proteins) for cancer therapy.
ASSOCIATED CONTENT
*
SSupporting Information
The Supporting Information is available free of charge on the
ACS Publications website at DOI: 10.1021/acs.nano-
lett.9b01660.
Materials and methods, synthesis and characterization of
the SA-MTO prodrug, polymers and NPs, body weight
of mice, histology, and immune response; NMR and
uorescence emission spectra, acidbase titration
prole, size distribution prole, CLSM images, PLK1
expression, cell viability and body weight graphs,
uorescence image of the tumors and main organs,
histological section of the major organs, serum level
graphs (PDF)
AUTHOR INFORMATION
Corresponding Authors
*E-mail: ofarokhzad@bwh.harvard.edu.
*E-mail: xuxiaod5@mail.sysu.edu.cn.
ORCID
Wei Tao: 0000-0002-4277-3728
Omid C Farokhzad: 0000-0003-2009-270X
Xiaoding Xu: 0000-0002-9785-6731
Author Contributions
P.E.S., H.Y., and C.L. contributed equally to this work.
Notes
Theauthorsdeclarethefollowingcompetingnancial
interest(s): O.C.F. has nancial interests in Selecta Bio-
sciences, Tarveda Therapeutics, Placon Therapeutics, and Seer.
ACKNOWLEDGMENTS
This work was supported by the National Natural Science
Foundation of China (81874226 and 81803020), the Interna-
tional Scientic and Technological Cooperation Program from
Guangdong Science and Technology Department
(2018A050506033), the Thousand Talents Program for
Distinguished Young Scholars, the grants from Guangzhou
Science and Technology Bureau (201902020015 and
201704020131) and Guangdong Science and Technology
Department (2017B030314026), the Three million for Three
YearsProject of the High-level Talent Special Funding
Scheme of Sun Yat-Sen Memorial Hospital, the David H.
Koch-Prostate Cancer Foundation (PCF) Program in Cancer
Nanotherapeutics, and the US METAvivor Early Career
Investigator Award (2018A020560).
REFERENCES
(1) Fire, A.; Xu, S.; Montgomery, M. K.; Kostas, S. A.; Driver, S. E.;
Mello, C. C. Nature 1998,391, 806811.
(2) Elbashir, S. M.; Harborth, J.; Lendeckel, W.; Yalcin, A.; Weber,
K.; Tuschl, T. Nature 2001,411, 494498.
(3) McCaffrey, A. P.; Meuse, L.; Pham, T.-T. T.; Conklin, D. S.;
Hannon, G. J.; Kay, M. A. Nature 2002,418,3839.
(4) Whitehead, K. A.; Langer, R.; Anderson, D. G. Nat. Rev. Drug
Discovery 2009,8, 129138.
(5) Yin, H.; Kanasty, R. L.; Eltoukhy, A. A.; Vegas, A. J.; Dorkin, J.
R.; Anderson, D. G. Nat. Rev. Genet. 2014,15, 541555.
(6) Shi, J.; Kantoff, P. W.; Wooster, R.; Farokhzad, O. C. Nat. Rev.
Cancer 2017,17,2037.
(7) Tseng, Y. C.; Mozumdar, S.; Huang, L. Adv. Drug Delivery Rev.
2009,61, 721731.
(8) Howard, K. A. Adv. Drug Delivery Rev. 2009,61, 710720.
(9) Zuckerman, J. E.; Davis, M. E. Nat. Rev. Drug Discovery 2015,14,
843856.
(10) Liu, Y.; Xu, C. F.; Iqbal, S.; Yang, X. Z.; Wang, J. Adv. Drug
Delivery Rev. 2017,115,98114.
(11) Kanasty, R.; Dorkin, J. R.; Vegas, A.; Anderson, D. Nat. Mater.
2013,12, 967977.
(12) Lu, Y.; Aimetti, A. A.; Langer, R.; Gu, Z. Nat. Rev. Mater. 2017,
2, 16075.
(13) Sun, C.-Y.; Shen, S.; Xu, C.-F.; Li, H.-J.; Liu, Y.; Cao, Z.-T.;
Yang, X.-Z.; Xia, J.-X.; Wang, J. J. Am. Chem. Soc. 2015,137, 15217
15224.
(14) Xu, X.; Wu, J.; Liu, Y.; Yu, M.; Zhao, L.; Zhu, X.; Bhasin, S.; Li,
Q.; Ha, E.; Shi, J.; Farokhzad, O. C. Angew. Chem., Int. Ed. 2016,55,
70917094.
(15) Xu, X.; Wu, J.; Liu, Y.; Saw, P. E.; Tao, W.; Yu, M.; Zope, H.;
Si, M.; Victorious, A.; Rasmussen, J.; Ayyash, D.; Farokhzad, O. C.;
Shi, J. ACS Nano 2017,11, 26182627.
Nano Letters Letter
DOI: 10.1021/acs.nanolett.9b01660
Nano Lett. 2019, 19, 59675974
5973
(16) Yang, X.-Z.; Du, J.-Z.; Dou, S.; Mao, C.-Q.; Long, H.-Y.; Wang,
J. ACS Nano 2012,6, 771781.
(17) Zhou, Z.; Liu, X.; Zhu, D.; Wang, Y.; Zhang, Z.; Zhou, X.; Qiu,
N.; Chen, X.; Shen, Y. Adv. Drug Delivery Rev. 2017,115, 115154.
(18) Liu, X.; Xiang, J.; Zhu, D.; Jiang, L.; Zhou, Z.; Tang, J.; Liu, X.;
Huang, Y.; Shen, Y. Adv. Mater. 2016,28, 17431752.
(19) Yu, H.; Zou, Y.; Wang, Y.; Huang, X.; Huang, G.; Sumer, B. D.;
Boothman, D. A.; Gao, J. ACS Nano 2011,5, 92469255.
(20) Bolcato-Bellemin, A. L.; Bonnet, M. E.; Creusat, G.; Erbacher,
P.; Behr, J. P. Proc. Natl. Acad. Sci. U. S. A. 2007,104, 1605016055.
(21) Choi, K. Y.; Silvestre, O. F.; Huang, X.; Hida, N.; Liu, G.; Ho,
D. N.; Lee, S.; Lee, S. W.; Hong, J. I.; Chen, X. Nat. Protoc. 2014,9,
19001915.
(22) Choi, K. Y.; Silvestre, O. F.; Huang, X.; Min, K. H.; Howard, G.
P.; Hida, N.; Jin, A. J.; Carvajal, N.; Lee, S. W.; Hong, J. I.; Chen, X.
ACS Nano 2014,8, 45594570.
(23) Liu, G.; Choi, K. Y.; Bhirde, A.; Swierczewska, M.; Yin, J.; Lee,
S. W.; Park, J. H.; Hong, J. I.; Xie, J.; Niu, G.; Kiesewetter, D. O.; Lee,
S.; Chen, X. Angew. Chem., Int. Ed. 2012,51, 445449.
(24) Min, K. H.; Kim, Y. H.; Wang, Z.; Kim, J.; Kim, J. S.; Kim, S.
H.; Kim, K.; Kwon, I. C.; Kiesewetter, D. O.; Chen, X. Theranostics
2017,7, 42404254.
(25) Vargas, J. R.; Stanzl, E. G.; Teng, N. N. H.; Wender, P. A. Mol.
Pharmaceutics 2014,11, 25532565.
(26) Ren, Y.; Cheung, H. W.; von Maltzhan, G.; Agrawal, A.;
Cowley, G. S.; Weir, B. A.; Boehm, J. S.; Tamayo, P.; Karst, A. M.;
Liu, J. F.; Hirsch, M. S.; Mesirov, J. P.; Drapkin, R.; Root, D. E.; Lo, J.;
Fogal, V.; Ruoslahti, E.; Hahn, W. C.; Bhatia, S. N. Sci. Transl. Med.
2012,4, 147ra112.
(27) Mitchell, D. J.; Steinman, L.; Kim, D. T.; Fathman, C. G.;
Rothbard, J. B. J. Pept. Res. 2000,56, 318325.
(28) Zhao, Y.; Wang, W.; Guo, S.; Wang, Y.; Miao, L.; Xiong, Y.;
Huang, L. Nat. Commun. 2016,7, 11822.
(29) Zou, Y.; Zheng, M.; Yang, W.; Meng, F.; Miyata, K.; Kim, H. J.;
Kataoka, K.; Zhong, Z. Adv. Mater. 2017,29, 1703285.
(30) Li, J.; Yu, X.; Wang, Y.; Yuan, Y.; Xiao, H.; Cheng, D.; Shuai, X.
Adv. Mater. 2014,26, 82178224.
(31) Xu, X.; Wu, J.; Liu, S.; Saw, P. E.; Tao, W.; Li, Y.; Krygsman, L.;
Yegnasubramanian, S.; De Marzo, A. M.; Shi, J.; Bieberich, C. J.;
Farokhzad, O. C. Small 2018,14, 1802565.
(32) Chen, W.; Meng, F.; Cheng, R.; Deng, C.; Feijen, J.; Zhong, Z.
J. Controlled Release 2014,190, 398414.
(33) Bildstein, L.; Dubernet, C.; Couvreur, P. Adv. Drug Delivery Rev.
2011,63,323.
(34) Sinhababu, A. K.; Thakker, D. R. Adv. Drug Delivery Rev. 1996,
19, 241273.
(35) Mahato, R.; Tai, W.; Cheng, K. Adv. Drug Delivery Rev. 2011,
63, 659670.
(36) Stella, V. J.; Nti-Addae, K. W. Adv. Drug Delivery Rev. 2007,59,
677694.
(37) Xu, X. D.; Cheng, Y. J.; Wu, J.; Cheng, H.; Cheng, S. X.; Zhuo,
R. X.; Zhang, X. Z. Biomaterials 2016,76, 238249.
(38) Chang, R. S.; Suh, M. S.; Kim, S.; Shim, G.; Lee, S.; Han, S. S.;
Lee, K. E.; Jeon, H.; Choi, H.-G.; Choi, Y.; Kim, C.-W.; Oh, Y.-K.
Biomaterials 2011,32, 97859795.
(39) Pernicova, I.; Korbonits, M. Nat. Rev. Endocrinol. 2014,10,
143156.
(40) Zhou, K.; Wang, Y.; Huang, X.; Luby-Phelps, K.; Sumer, B. D.;
Gao, J. Angew. Chem., Int. Ed. 2011,50, 61096114.
(41) Wang, Y.; Zhou, K.; Huang, G.; Hensley, C.; Huang, X.; Ma,
X.; Zhao, T.; Sumer, B. D.; DeBerardinis, R. J.; Gao, J. Nat. Mater.
2014,13, 204212.
(42) Yang, Z. M.; Xu, K. M.; Guo, Z. F.; Guo, Z. H.; Xu, B. Adv.
Mater. 2007,19, 31523156.
(43) McGoldrick, C. A.; Jiang, Y.-L.; Paromov, V.; Brannon, M.;
Krishnan, K.; Stone, W. L. BMC Cancer 2014,14, 77.
(44) McGoldrick, C. A.; Jiang, Y.-L.; Brannon, M.; Krishnan, K.;
Stone, W. L. BMC Cancer 2014,14, 675.
(45) Takai, N.; Hamanaka, R.; Yoshimatsu, J.; Miyakawa, I. Oncogene
2005,24, 287291.
(46) Strebhardt, K. Nat. Rev. Drug Discovery 2010,9, 643660.
(47) Strebhardt, K.; Ullrich, A. Nat. Rev. Cancer 2006,6, 321330.
(48) Gjertsen, B. T.; Schöffski, P. Leukemia 2015,29,1119.
(49) Liu, Z.; Sun, Q.; Wang, X. Transl Oncol 2017,10,2232.
(50) de Cá
rcer, G.; Venkateswaran, S. V.; Salgueiro, L.; El Bakkali,
A.; Somogyi, K.; Rowald, K.; Montañé
s, P.; Sanclemente, M.; Escobar,
B.; de Martino, A.; McGranahan, N.; Malumbres, M.; Sotillo, R. Nat.
Commun. 2018,9, 3012.
(51) Liu, X. Transl Oncol 2015,8, 185195.
(52) Gutteridge, R. E.; Ndiaye, M. A.; Liu, X.; Ahmad, N. Mol.
Cancer Ther. 2016,15, 14271435.
(53) Song, B.; Liu, X. S.; Rice, S. J.; Kuang, S.; Elzey, B. D.;
Konieczny, S. F.; Ratliff, T. L.; Hazbun, T.; Chiorean, E. G.; Liu, X.
Mol. Cancer Ther. 2013,12,5868.
(54) Liu, X. S.; Song, B.; Tang, J.; Liu, W.; Kuang, S.; Liu, X. Mol.
Cancer Ther. 2012,32, 40534067.
(55) DAmico, A. V. J. Clin. Oncol. 2014,32, 362364.
(56) Bell, D. H. Biochim. Biophys. Acta, Gene Struct. Expression 1988,
949, 132137.
(57) Liu, P.; Qin, L.; Wang, Q.; Sun, Y.; Zhu, M.; Shen, M.; Duan, Y.
Biomaterials 2012,33, 67396747.
(58) Xu, X.; Saw, P. E.; Tao, W.; Li, Y.; Ji, X.; Bhasin, S.; Liu, Y.;
Ayyash, D.; Rasmussen, J.; Huo, M.; Shi, J.; Farokhzad, O. C. Adv.
Mater. 2017,29, 1700141.
(59) Zhu, X.; Xu, Y.; Solis, L. M.; Tao, W.; Wang, L.; Behrens, C.;
Xu, X.; Zhao, L.; Liu, D.; Wu, J.; Zhang, N.; Wistuba, I. I.; Farokhzad,
O. C.; Zetter, B. R.; Shi, J. Proc. Natl. Acad. Sci. U. S. A. 2015,112,
77797784.
(60) Xu, X.; Xie, K.; Zhang, X.-Q.; Pridgen, E. M.; Park, G. Y.; Cui,
D. S.; Shi, J.; Wu, J.; Kantoff, P. W.; Lippard, S. J.; Langer, R.; Walker,
G. C.; Farokhzad, O. C. Proc. Natl. Acad. Sci. U. S. A. 2013,110,
1863818643.
(61) Liu, Y.; Gunda, V.; Zhu, X.; Xu, X.; Wu, J.; Askhatova, D.;
Farokhzad, O. C.; Parangi, S.; Shi, J. Proc. Natl. Acad. Sci. U. S. A.
2016,113, 77507755.
(62) Kim, T.; Afonin, K. A.; Viard, M.; Koyfman, A. Y.; Sparks, S.;
Heldman, E.; Grinberg, S.; Linder, C.; Blumenthal, R. P.; Shapiro, B.
A. Mol. Ther.Nucleic Acids 2013,2, e80.
(63) Xu, X.; Saw, P. E.; Tao, W.; Li, Y.; Ji, X.; Yu, M.; Mahmoudi,
M.; Rasmussen, J.; Ayyash, D.; Zhou, Y.; Farokhzad, O. C.; Shi, J.
Nano Lett. 2017,17, 44274435.
(64) Suk, J. S.; Xu, Q.; Kim, N.; Hanes, J.; Ensign, L. M. Adv. Drug
Delivery Rev. 2016,99,2851.
(65) Knop, K.; Hoogenboom, R.; Fischer, D.; Schubert, U. S. Angew.
Chem., Int. Ed. 2010,49, 62886308.
(66) Bertrand, N.; Wu, J.; Xu, X.; Kamaly, N.; Farokhzad, O. C. Adv.
Drug Delivery Rev. 2014,66,225.
(67) Li, H.-J.; Du, J.-Z.; Liu, J.; Du, X.-J.; Shen, S.; Zhu, Y.-H.; Wang,
X.; Ye, X.; Nie, S.; Wang, J. ACS Nano 2016,10, 67536761.
(68) Li, H.-J.; Du, J.-Z.; Du, X.-J.; Xu, C.-F.; Sun, C.-Y.; Wang, H.-
X.; Cao, Z.-T.; Yang, X.-Z.; Zhu, Y.-H.; Nie, S.; Wang, J. Proc. Natl.
Acad. Sci. U. S. A. 2016,113, 41644169.
(69) Sugahara, K. N.; Teesalu, T.; Karmali, P. P.; Kotamraju, V. R.;
Agemy, L.; Girard, O. M.; Hanahan, D.; Mattrey, R. F.; Ruoslahti, E.
Cancer Cell 2009,16, 510520.
Nano Letters Letter
DOI: 10.1021/acs.nanolett.9b01660
Nano Lett. 2019, 19, 59675974
5974
... As compared to nanoparticle-mediated, noncovalent drug encapsulation, stimulusresponsive prodrug nanoparticles have a pharmaceutical advantage: they can be tuned to minimize drug leakage and to control drug release profiles through chemical linkers 209,210 and have recently been developed as a strategy to enhance the cancer immunotherapy effectiveness. 200 Iron oxide-zinc oxide core-shell NPs have been successfully used to deliver carcinoembryonic antigen into dendritic cells while simultaneously acting as an imaging agent. ...
Preprint
Full-text available
Rapid and sustained growth in the field of immuno-oncology has resulted in expansion of available scientific literature. Gaining valuable insights and establishing deep and often hidden meaningful connections in such a large body of work is the need of the hour. In this report we summarize our findings from a novel Natural Language Programming (NLP)-based approach on a large dataset of >350K scientific publications in immuno-oncology research spanning across two decades (2000-2022) retrieved from the CAS Content Collection. Our analysis led to identification of >300 emerging concepts across major categories such as therapeutic targets, biomarkers, therapies, and types of cancer. We present a “Trend Landscape Map” of emerging concepts in immuno-oncology possessing layers of intricacies – at first glance providing information for the >300 identified concepts arranged hierarchically across 8 major categories and at a deeper level providing detailed quantitative metrics of growth over the last three years (2020-2022). While concepts such as immune checkpoint inhibitors (ICIs), antibody-drug conjugates (ADCs) and chimeric antigenic receptors (CARs) continue to be important in immuno-oncology, their growth over the last three years have been modest. On the other hand, concepts including protein targets such as TROP2, nectin-4, and gasdermins display rapid increase in scientific publications over 2020-2022 while their absolute number of publications remain low potentially indicative of early emergence. Finally, guided by our trend landscape analysis, we performed substance data analysis leveraging data from >3.2 million substances from the CAS Registry and identified potential higher commercial interest in protein/peptide sequences rather than small molecules in cancer immunotherapy as seen with respect to patent publications. It is our hope that our subject matter experts' knowledge-guided big data analysis approach based on the corpus of robustly CAS indexed data provides a comprehensive picture of immuno-oncology as it stands today with the trend landscape map serving as a valuable resource to researchers in this field.
... Since the positively charged lipids can form stable complexes with negatively charged nucleic acids through electrostatic interactions, these complexes can not only prevent the RNA molecules from being degraded but also enhance their intracellular transfection efficiency. [15] For instance, Mitchell et al. proposed a co-assembled delivery approach for doxorubicin (DOX) and siRNA, which synergistically combines the chemotherapy and RNA interference together by conjugating DOX onto the surface of LNPs loaded with Bcl-2 gene-targeted siRNA, resulting in robust suppression of Bcl-2 expression in Burkitt's lymphoma cells and effective tumor treatment. [16] Despite considerable success of LNPs in siRNA delivery for targeted gene therapy, the low efficiency of siRNA entry into the cytosol remains a fundamental challenge. ...
Article
Full-text available
Ribonucleic acid (RNA) plays a pivotal role in gene regulation and protein biosynthesis. Interfering the physiological function of key RNAs to induce cell apoptosis holds great promise for cancer treatment. Many RNA‐targeted anti‐cancer strategies have emerged continuously. Among them, RNA interference (RNAi) has been recognized as a promising therapeutic modality for various disease treatments. Nevertheless, the primary obstacle in siRNA delivery‐escaping the endosome and crossing the plasma membrane severely impedes its therapeutic potential. Thus far, a variety of nanosystems as well as carrier‐free bioconjugation for siRNA delivery have been developed and employed to enhance the drug delivery and anti‐tumor efficiency. Besides, the use of small molecules to target specific RNA structures and disrupt their function, along with the covalent modification of RNA, has also drawn tremendous attention recently owing to high therapeutic efficacy. In this review, we will provide an overview of recent progress in RNA‐targeted cancer therapy including various siRNA delivery strategies, RNA‐targeting small molecules, and newly emerged covalent RNA modification. Finally, challenges and future perspectives faced in this research field will be discussed.
Article
Full-text available
Fundus neovascularization diseases are a series of blinding eye diseases that seriously impair vision worldwide. Currently, the means of treating these diseases in clinical practice are continuously evolving and have rapidly revolutionized treatment opinions. However, key issues such as inadequate treatment effectiveness, high rates of recurrence, and poor patient compliance still need to be urgently addressed. Multifunctional nanomedicine can specifically respond to both endogenous and exogenous microenvironments, effectively deliver drugs to specific targets and participate in activities such as biological imaging and the detection of small molecules. Nano-in-micro (NIM) delivery systems such as metal, metal oxide and up-conversion nanoparticles (NPs), quantum dots, and carbon materials, have shown certain advantages in overcoming the presence of physiological barriers within the eyeball and are widely used in the treatment of ophthalmic diseases. Few studies, however, have evaluated the efficacy of NIM delivery systems in treating fundus neovascular diseases (FNDs). The present study describes the main clinical treatment strategies and the adverse events associated with the treatment of FNDs with NIM delivery systems and summarizes the anatomical obstacles that must be overcome. In this review, we wish to highlight the principle of intraocular microenvironment normalization, aiming to provide a more rational approach for designing new NIM delivery systems to treat specific FNDs. Graphical Abstract
Article
Ribonucleic acid (RNA) therapeutics offer a broad prospect in cancer treatment. However, their successful application requires overcoming various physiological barriers to effectively deliver RNAs to the target sites. Currently, a number of RNA delivery systems based on polymeric nanoparticles are developed to overcome these barriers in RNA delivery. This work provides an overview of the existing RNA therapeutics for cancer gene therapy, and particularly summarizes those that are entering the clinical phase. This work then discusses the core features and latest research developments of tumor microenvironment‐responsive polymer‐based RNA delivery carriers which are designed based on the pathological characteristics of the tumor microenvironment. Finally, this work also proposes opportunities for the transformation of RNA therapies into cancer immunotherapy methods in clinical applications.
Article
In the ever-evolving landscape of cancer research, immuno-oncology stands as a beacon of hope, offering novel avenues for treatment. This study capitalizes on the vast repository of immuno-oncology-related scientific documents within the CAS Content Collection, totaling over 350,000, encompassing journals and patents. Through a pioneering approach melding natural language processing with the CAS indexing system, we unveil over 300 emerging concepts, depicted in a comprehensive “Trend Landscape Map”. These concepts, spanning therapeutic targets, biomarkers, and types of cancers among others, are hierarchically organized into eight major categories. Delving deeper, our analysis furnishes detailed quantitative metrics showcasing growth trends over the past three years. Our findings not only provide valuable insights for guiding future research endeavors but also underscore the merit of tapping the vast and unparalleled breadth of existing scientific information to derive profound insights.
Article
Gene therapy uses modern molecular biology methods to repair disease‐causing genes. As a burgeoning therapeutic, it has been widely applied for cancer therapy. Since 1989, there have been numerous clinical gene therapy cases worldwide. However, a few are successful. The main challenge of clinical gene therapy is the lack of efficient and safe vectors. Although viral vectors show high transfection efficiency, their application is still limited by immune rejection and packaging capacity. Therefore, the development of non‐viral vectors is overwhelming. Nanoplatform‐based non‐viral vectors become a hotspot in gene therapy. The reasons are mainly as follows. 1) Non‐viral vectors can be engineered to be uptaken by specific types of cells or tissues, providing effective targeting capability. 2) Non‐viral vectors can protect goods that need to be delivered from degradation. 3) Nanoparticles can transport large‐sized cargo such as CRISPR/Cas9 plasmids and nucleoprotein complexes. 4) Nanoparticles are highly biosafe, and they are not mutagenic in themselves compared to viral vectors. 5) Nanoparticles are easy to scale preparation, which is conducive to clinical conversion and application. Here, an overview of the categories of nanoplatform‐based non‐viral gene vectors, the limitations on their development, and their applications in cancer therapy.
Article
RNA interference (RNAi) technology has been a promising treatment strategy for combating intractable diseases. However, the applications of RNAi in clinical were hampered by extracellular and intracellular barriers. To overcome these barriers, various siRNA delivery systems have been developed in the past two decades. The first approved RNAi therapeutic, Patisiran (ONPATTRO™) using lipids as the carrier, for the treatment of amyloidosis is one of the most important milestones. This has greatly encouraged researchers to work on creating new functional siRNA carriers. In this review, we summarized the recent advances in siRNA carriers consisting of lipids, polymers, and polymer‐modified inorganic particles for cancer therapy. Representative examples are presented to show the structural design of the carriers in order to overcome the delivery hurdles associated with RNAi therapies. Finally, the existing challenges and future perspective for developing RNAi as a clinical modality will be discussed and proposed. We believe that the addressed contributions in this review will promote the development of siRNA delivery systems for future clinical applications. This article is protected by copyright. All rights reserved
Article
Full-text available
Emerging evidence has demonstrated the significant contribution of mitochondrial metabolism dysfunction to promote cancer development and progression. Aberrant expression of mitochondrial genome (mtDNA)‐encoded proteins widely involves mitochondrial metabolism dysfunction, and targeted regulation of their expression can be an effective strategy for cancer therapy, which however is challenged due to the protection by the mitochondrial double membrane. Herein, a mitochondria‐targeted RNAi nanoparticle (NP) platform for effective regulation of mitochondrial metabolism and breast cancer (BCa) therapy is developed. This nanoplatform is composed of a hydrophilic polyethylene glycol (PEG) shell, a hydrophobic poly(2‐(diisopropylamino)ethyl methacrylate) (PDPA) core, and charged‐mediated complexes of mitochondria‐targeting and membrane‐penetrating peptide amphiphile (MMPA) and small interfering RNA (siRNA) embedded in the core. After tumor accumulation and internalization by tumor cells, these NPs can respond to the endosomal pH to expose the MMPA/siRNA complexes, which can specifically transport siRNA into the mitochondria to down‐regulate mtDNA‐encoded protein expression (e.g., ATP6 and CYB). More importantly, because ATP6 down‐regulation can suppress ATP production and enhance reactive oxygen species (ROS) generation to induce mitochondrial damage and mtDNA leakage into tumor tissues, the NPs can combinatorially inhibit tumor growth via suppressing ATP production and repolarizing tumor‐associated macrophages (TAMs) into tumor‐inhibiting M1‐like macrophages by mtDNA.
Article
Full-text available
Polo-like kinase 1 (Plk1) is overexpressed in a wide spectrum of human tumors, being frequently considered as an oncogene and an attractive cancer target. However, its contribution to tumor development is unclear. Using a new inducible knock-in mouse model we report here that Plk1 overexpression results in abnormal chromosome segregation and cytokinesis, generating polyploid cells with reduced proliferative potential. Mechanistically, these cyto-kinesis defects correlate with defective loading of Cep55 and ESCRT complexes to the abscission bridge, in a Plk1 kinase-dependent manner. In vivo, Plk1 overexpression prevents the development of Kras-induced and Her2-induced mammary gland tumors, in the presence of increased rates of chromosome instability. In patients, Plk1 overexpression correlates with improved survival in specific breast cancer subtypes. Therefore, despite the therapeutic benefits of inhibiting Plk1 due to its essential role in tumor cell cycles, Plk1 overexpression has tumor-suppressive properties by perturbing mitotic progression and cytokinesis.
Article
Full-text available
Combination cancer treatment has emerged as a critical approach to achieve remarkable anticancer effect. In this study, we prepared a theranostic nanoformulation that allows for photoacoustic imaging as well as combination gene and photothermal therapy. Gold nanorods (GNR) were coated with dipicolyl amine (DPA), which forms stable complexes with Zn²⁺ cations. The resulting nanoparticles, Zn(II)/DPA-GNR, recognize phosphate-containing molecules, including siRNA, because of the specific interaction between Zn(II) and the phosphates. We chose anti-polo-like kinase 1 siRNA (siPLK) as our example for gene silencing. The strong complexation between Zn(II)/DPA-GNR and siPLK provided high stability to the nano-complexes, which efficiently delivered siRNA into the targeted cancer cells in vitro and in vivo. The particle served as a theranostic agent because the GNRs of nano-complexes permitted effective photothermal therapy as well as photoacoustic imaging upon laser irradiation. This gene/photothermal combination therapy using siPLK/Zn(II)DPA-GNRs exhibited significant antitumor activity in a PC-3 tumor mouse model. The concept described in this work may be extended to the development of efficient delivery strategies for other polynucleotides as well as advanced anticancer therapy.
Article
Full-text available
With the capability of specific silencing of target gene expression, RNA interference (RNAi) technology is emerging as a promising therapeutic modality for the treatment of cancer and other diseases. One key challenge for the clinical applications of RNAi is the safe and effective delivery of RNAi agents such as small interfering RNA (siRNA) to a particular non-liver diseased tissue (e.g., tumor) and cell type with sufficient cytosolic transport. In this work, we proposed a multifunctional envelope-type nanoparticle (NP) platform for prostate cancer (PCa)-specific in vivo siRNA delivery. A library of oligoarginine-functionalized and sharp pH-responsive polymers were synthesized and used for self-assembly with siRNA into NPs with the features of long blood circulation and pH-triggered oligoarginine-mediated endosomal membrane penetration. By further modification with ACUPA, a small molecular ligand specifically recognizing prostate-specific membrane antigen (PSMA) receptor, this envelope-type nanoplatform with multifunctional properties can efficiently target PSMA-expressing PCa cells and silence target gene expression. Systemic delivery of the siRNA NPs can efficiently silence the expression of prohibitin1 (PHB1), which is upregulated in PCa and other cancers, and significantly inhibit PCa tumor growth. These results suggest that this multifunctional envelope-type nanoplatform could become an effective tool for PCa-specific therapy.
Article
Full-text available
Polo-like kinase 1 (PLK1) plays an important role in the initiation, maintenance, and completion of mitosis. Dysfunction of PLK1 may promote cancerous transformation and drive its progression. PLK1 overexpression has been found in a variety of human cancers and was associated with poor prognoses in cancers. Many studies have showed that inhibition of PLK1 could lead to death of cancer cells by interfering with multiple stages of mitosis. Thus, PLK1 is expected to be a potential target for cancer therapy. In this article, we examined PLK1’s structural characteristics, its regulatory roles in cell mitosis, PLK1 expression, and its association with survival prognoses of cancer patients in a wide variety of cancer types, PLK1 interaction networks, and PLK1 inhibitors under investigation. Finally, we discussed the key issues in the development of PLK1-targeted cancer therapy.
Article
Biodegradable polymeric nanoparticles (NPs) have demonstrated significant potential to improve the systemic delivery of RNA interference (RNAi) therapeutics, such as small interfering RNA (siRNA), for cancer therapy. However, the slow and inefficient siRNA release inside tumor cells generally observed for most biodegradable polymeric NPs may result in compromised gene silencing efficacy. Herein, a biodegradable and redox‐responsive NP platform, composed of a solid poly(disulfide amide) (PDSA)/cationic lipid core and a lipid–poly(ethylene glycol) (lipid–PEG) shell for systemic siRNA delivery to tumor cells, is developed. This newly generated NP platform can efficiently encapsulate siRNA under extracellular environments and can respond to the highly concentrated glutathione (GSH) in the cytoplasm to induce fast intracellular siRNA release. By screening a library of PDSA polymers with different structures and chain lengths, the optimized NP platform shows the unique features of i) long blood circulation, ii) high tumor accumulation, iii) fast GSH‐triggered intracellular siRNA release, and iv) exceptionally effective gene silencing. Together with the facile polymer synthesis technique and robust NP formulation enabling scale‐up, this new redox‐responsive NP platform may become an effective tool for RNAi‐based cancer therapy.
Article
Small interfering RNA (siRNA) offers a highly selective and effective pharmaceutical for various life-threatening diseases, including cancers. The clinical translation of siRNA is, however, challenged by its short plasma life, poor cell uptake, and cumbersome intracellular trafficking. Here, cNGQGEQc peptide-functionalized reversibly crosslinked chimaeric polymersomes (cNGQ/RCCPs) is shown to mediate high-efficiency targeted delivery of Polo-like kinase1 specific siRNA (siPLK1) to orthotopic human lung cancer in nude mice. Strikingly, siRNA is completely and tightly loaded into the aqueous lumen of the polymersomes at an unprecedentedly low N/P ratio of 0.45. cNGQ/RCCPs loaded with firefly luciferase specific siRNA (siGL3) or siPLK1 are efficiently taken up by α3β1-integrin-overexpressing A549 lung cancer cells and quickly release the payloads to the cytoplasm, inducing highly potent and sequence-specific gene silencing in vitro. The in vivo studies using nude mice bearing orthotopic A549 human lung tumors reveal that siPLK1-loaded cNGQ/RCCPs boost long circulation, superb tumor accumulation and selectivity, effective suppression of tumor growth, and significantly improved survival time. These virus-mimicking chimaeric polymersomes provide a robust and potent platform for targeted cancer siRNA therapy.
Article
Gene therapy represents a promising cancer treatment featuring high efficacy and limited side effects, but it is stymied by a lack of safe and efficient gene-delivery vectors. Cationic polymers and lipid-based nonviral gene vectors have many advantages and have been extensively explored for cancer gene delivery, but their low gene-expression efficiencies relative to viral vectors limit their clinical translations. Great efforts have thus been devoted to developing new carrier materials and fabricating functional vectors aimed at improving gene expression, but the overall efficiencies are still more or less at the same level. This review analyzes the cancer gene-delivery cascade and the barriers, the needed nanoproperties and the current strategies for overcoming these barriers, and outlines PEGylation, surface-charge, size, and stability dilemmas in vector nanoproperties to efficiently accomplish the cancer gene-delivery cascade. Stability, surface, and size transitions (3S Transitions) are proposed to resolve those dilemmas and strategies to realize these transitions are comprehensively summarized. The review concludes with a discussion of the future research directions to design high-performance nonviral gene vectors.
Article
The application of nanoparticles (NPs) to drug delivery has led to the development of novel nanotherapeutics for the treatment of various diseases including cancer. However, clinical use of NP-mediated drug delivery has not always translated into improved survival of cancer patients, in part due to the suboptimal properties of NP platforms, such as premature drug leakage during preparation, storage, or blood circulation, lack of active targeting to tumor tissue and cells, and poor tissue penetration. Herein, an innovative reactive oxygen species (ROS)-responsive polyprodrug is reported that can self-assemble into stable NPs with high drug loading. This new NP platform is composed of the following key components: (i) polyprodrug inner core that can respond to ROS for triggered release of intact therapeutic molecules, (ii) polyethylene glycol (PEG) outer shell to prolong blood circulation; and (iii) surface-encoded internalizing RGD (iRGD) to enhance tumor targeting and tissue penetration. These targeted ROS-responsive polyprodrug NPs show significant inhibition of tumor cell growth both in vitro and in vivo.
Article
While RNA interference (RNAi) therapy has demonstrated significant potential for cancer treatment, effective and safe systemic delivery of RNAi agents such as small interfering RNA (siRNA) into tumor cells in vivo remains challenging. We herein reported a unique multistaged siRNA delivery nanoparticle (NP) platform, which is comprised of (i) a polyethylene glycol (PEG) surface shell, (ii) a sharp tumor microenvironment (TME) pH-responsive polymer that forms the NP core, and (iii) charge-mediated complexes of siRNA and tumor cell-targeting- and penetrating-peptide-amphiphile (TCPA) that are encapsulated in the NP core. When the rationally designed, long circulating polymeric NPs accumulate in tumor tissues after intravenous administration, the targeted siRNA-TCPA complexes can be rapidly released via TME pH-mediated NP disassembly for subsequent specific targeting of tumor cells and cytosolic transport, thus achieving efficient gene silencing. In vivo results further demonstrate that the multistaged NP delivery of siRNA against bromodomain 4 (BRD4), a recently discovered target protein that regulates the development and progression of prostate cancer (PCa), can significantly inhibit PCa tumor growth.
Article
Cascades of systemic and intracellular obstacles, including low stability in blood, little tumor accumulation, weak tumor penetration, poor cellular uptake, inefficient endosomal escape and deficient disassembly in the cytoplasm, must be overcome in order to deliver nucleic acid drugs for cancer therapy. Nanocarriers that are sensitive to a variety of physiological stimuli, such as pH, redox status, and cell enzymes, are substantially changing the landscape of nucleic acid drug delivery by helping to overcome cascaded systemic and intracellular barriers. This review discusses nucleic acid-based therapeutics, systemic and intracellular barriers to efficient nucleic acid delivery, and nanocarriers responsive to extracellular and intracellular biological stimuli to overcome individual barriers. In particular, responsive nanocarriers for the cascaded delivery of nucleic acids in vivo are highlighted. Developing novel cascaded nanocarriers that transform their physicochemical properties in response to various stimuli in a timely and spatially controlled manner for nucleic acid drug delivery holds great potential for translating the promise of nucleic acid drugs and achieving clinically successful cancer therapy.